Elsevier

Acta Biomaterialia

Volume 8, Issue 7, July 2012, Pages 2815-2823
Acta Biomaterialia

Revision joint replacement, wear particles, and macrophage polarization

https://doi.org/10.1016/j.actbio.2012.03.042Get rights and content

Abstract

Currently, younger, more active patients are being offered total joint replacement (TJR) for end-stage arthritic disorders. Despite improved durability of TJRs, particle-associated wear of the bearing surfaces continues to be associated with particulate debris, which can activate monocyte/macrophages. Activated macrophages then produce pro-inflammatory factors and cytokines that induce an inflammatory reaction that activates osteoclasts leading to bone breakdown and aseptic loosening. We hypothesized that activated macrophages in tissues harvested from revised joint replacements predominantly express an M1 pro-inflammatory phenotype due to wear-particle-associated cell activation, rather than an M2 anti-inflammatory phenotype. We further questioned whether it is possible to convert uncommitted monocyte/macrophages to an M2 phenotype by the addition of interleukin-4 (IL-4), or whether it is necessary to first pass through an M1 intermediate stage. Retrieved periprosthetic tissues demonstrated increased M1/M2 macrophage ratios compared to non-operated osteoarthritic synovial tissues, using immunohistochemical staining and Western blotting. Uncommitted monocyte/macrophages with/without polymethyl-methacrylate particles were transformed to an M2 phenotype by IL-4 more efficiently when the cells were first passed through an M1 phenotype by exposure to endotoxin. Wear particles induce a pro-inflammatory microenvironment that facilitates osteolysis; these events may potentially be modulated favorably by exposure to IL-4.

Introduction

Total joint replacement (TJR) is a successful operation for patients suffering from disabling arthritis and other degenerative conditions. However, wear of artificial joints occurs in association with the level of activity and duration of implantation. TJR failure is often associated with osteolysis and is a long-term complication that may require revision surgery [1], [2], [3], [4], [5]. Production of wear particles which are biologically active and indigestible incites an innate inflammatory reaction that may lead to periprosthetic bone loss, implant loosening and pathological fracture through osteolytic bone [4], [5], [6], [7], [8]. Particles are phagocytosed by monocyte/macrophage lineage cells, leading to their proliferation, differentiation, and activation [1], [9], [10]. These events lead to intracellular signal transduction involving activation of transcription factor NFkB and nuclear translocation, which up-regulate gene expression mechanisms for pro-inflammatory cytokines, chemokines, and other substances [5], [11], [12]. The end result is the disruption of the homeostatic balance between bone formation and resorption [5], [11], [12].

A current hypothesis suggests that macrophage activation in osteolysis may culminate in a specific phenotype, with polarization to either M1 or M2 profile, due to the undifferentiated nature of monocyte/macrophage precursors, and the microenvironment of cell activation [9], [13]. Studies also suggest that there may be epigenetic control of macrophage polarization, suggesting a possible genetic predisposition for osteolysis [14], [15], [16]. Specifically, this hypothesis suggests that wear particles initiate the migration of monocyte/macrophage precursors to the local site of particle production, and subsequent differentiation and activation to a classical M1 phenotype that initially promotes acute inflammation. This acute inflammatory state overcomes the anti-inflammatory environment supported by alternatively activated M2 macrophages that normally promotes bone healing, debris scavenging, wound healing, and angiogenesis [11], [12].

The cytokine production profiles of M1 and M2 macrophages differ significantly and can be used to identify different predominant populations in a specific clinical situation. M1 macrophages produce primarily pro-inflammatory mediators, including TNF-α, IL-1, IL-6, and type 1 interferon, as well as IL-12 and IL-23, with the expression of inducible nitric oxide synthase (iNOS) and HLA-DR [17], [18], [19]. In contrast, M2 macrophages produce low levels of IL-12 and pro-inflammatory cytokines. The M2 profile is characterized by increased IL-4, IL-10, and IL-13 production, and expression of CCL1, CCL18, FIZZ1, mammalian chitinase Ym1, Arginase 1, CD163, and chitotriosidase [11], [12], [20], [21]. This differential cytokine production and receptor expression can be used to characterize which macrophages are present in a clinical situation. Once individual populations of M1 and M2 macrophages are identified, the cytokine profiles induced by pro-inflammatory stimuli such as lipopolysaccharide (LPS) or wear particles may be used to confirm the phenotype of the macrophages [22]. Lipopolysaccharide is a particularly relevant stimulus to joint replacement, as Greenfield and colleagues have demonstrated the presence of LPS in some retrieved tissues [23], [24], [25], [26].

The overall goal of this research was to identify macrophage populations in retrieved tissues from primary total joint replacement compared to revision surgeries with wear-particle-associated inflammation. First we hypothesized that there is a higher ratio of M1/M2 macrophages in tissues harvested from patients undergoing revision joint replacement compared to synovial tissues from patients undergoing primary joint replacement. We then questioned whether it was possible to isolate and modulate macrophage populations in vitro to selectively enhance an M2 profile in response to the inflammatory stimuli LPS and polymethyl-methacrylate (PMMA) particles by adding IL-4 to differentiate uncommitted macrophages towards a M2 phenotype [11]. We tested these hypotheses using immunohistochemistry, Western blot analysis, flow cytometry, and enzyme-linked immunosorbent (ELISA) assay of culture supernatants.

Section snippets

Tissue collection

This research was approved by the Stanford University School of Medicine’s Administrative Panel on Human Subjects in Medical Research. Synovium was collected during primary TJR and periprosthetic tissues during revision joint replacement. Periprosthetic tissues were obtained from patients with radiographic evidence of osteolysis who underwent revision total hip or knee arthroplasty in the absence of infection (all tissues were aerobic and anaerobic culture negative). We collected synovium from

Clinical investigation

The results are presented in the context of two experimental research plans. The first focuses on the clinical characterization of the synovium and pseudomembrane retrieval tissues by immunohistochemistry and Western blotting. Initial studies focused on the differential expression of M1 and M2 macrophages in human synovial tissue as compared to periprosthetic tissues.

Discussion

The purpose of this study was to determine whether wear particles associated with joint replacements influenced macrophage polarization. The experimental plan used human tissue retrievals and in vitro studies with isolated murine monocyte/macrophages. Although the revision surgery cases represent a variety of implant and wear debris types, which may be seen as a weakness of the study, as all cases demonstrated the same principles of macrophage polarization in response to wear debris, this gives

Acknowledgements

This research was supported by the Ellenburg Chair in Surgery at Stanford University and the Medical Scholars Program at Stanford University School of Medicine. We would like to thank Dr Nidhi Bhutani, Matt Decker, Subbus Dhulipala, and Andrew Peterman for their contributions.

References (57)

  • P.E. Purdue et al.

    The central role of wear debris in periprosthetic osteolysis

    Hss J

    (2006)
  • P. Koulouvaris et al.

    Expression profiling reveals alternative macrophage activation and impaired osteogenesis in periprosthetic osteolysis

    J Orthop Res

    (2008)
  • R. Chiu et al.

    Polymethylmethacrylate particles impair osteoprogenitor viability and expression of osteogenic transcription factors Runx2, osterix, and Dlx5

    J Orthop Res

    (2010)
  • R. Chiu et al.

    Ultrahigh molecular weight polyethylene wear debris inhibits osteoprogenitor proliferation and differentiation in vitro

    J Biomed Mater Res A

    (2009)
  • T. Ma et al.

    Biological effects of wear debris from joint arthroplasties

  • A. Marshall et al.

    How prevalent are implant wear and osteolysis, and how has the scope of osteolysis changed since 2000?

    J Am Acad Orthop Surg

    (2008)
  • P.H. Wooley et al.

    Aseptic loosening

    Gene Ther

    (2004)
  • P.E. Purdue

    Alternative macrophage activation in periprosthetic osteolysis

    Autoimmunity

    (2008)
  • V.W. Ho et al.

    Derivation and characterization of murine alternatively activated (M2) macrophages

    Methods Mol Biol

    (2009)
  • D.M. Mosser et al.

    Exploring the full spectrum of macrophage activation

    Nat Rev Immunol

    (2008)
  • O. Takeuch et al.

    Epigenetic control of macrophage polarization

    Eur J Immunol

    (2011)
  • J.M. Wilkinson et al.

    Variation in the TNF gene promoter and risk of osteolysis after total hip arthroplasty

    J Bone Miner Res

    (2003)
  • A. Gordon et al.

    Variation in the secreted frizzled-related protein-3 gene and risk of osteolysis and heterotopic ossification after total hip arthroplasty

    J Orthop Res

    (2007)
  • D.R. Sharda et al.

    Regulation of macrophage arginase expression and tumor growth by the ron receptor tyrosine kinase

    J Immunol

    (2011)
  • N. Kawanishi et al.

    Exercise training inhibits inflammation in adipose tissue via both suppression of macrophage infiltration and acceleration of phenotypic switching from M1 to M2 macrophages in high-fat-diet-induced obese mice

    Exerc Immunol Rev

    (2010)
  • C.N. Lumeng et al.

    Obesity induces a phenotypic switch in adipose tissue macrophage polarization

    J Clin Invest

    (2007)
  • G. Raes et al.

    FIZZ1 and Ym as tools to discriminate between differentially activated macrophages

    Dev Immunol

    (2002)
  • T.O. Jensen et al.

    Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma

    J Clin Oncol

    (2009)
  • Cited by (0)

    View full text